Brucella melitensis invades murine erythrocytes during infection

3 downloads 920 Views 1MB Size Report
Jul 7, 2014 - do not seem to multiply in erythrocytes and are found free in the cytoplasm. ..... AnalySIS and Adobe Photoshop software. 251 ..... could be infected during the course of infection by Brucella melitensis but strongly suggests.
IAI Accepts, published online ahead of print on 7 July 2014 Infect. Immun. doi:10.1128/IAI.01779-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Brucella melitensis invades murine erythrocytes during

2

infection

3 4

#

Marie-Alice Vitry, # Delphine Hanot Mambres, # Michaël Deghelt, #Katrin Hack, #Arnaud

5

Machelart, @Frédéric Lhomme, £Jean-Marie Vanderwinden, @Marjorie Vermeersch, *Carl De

6

Trez, @David Pérez-Morga, #,1 Jean-Jacques Letesson, †,1 Eric Muraille

7 8

#

9

Microbiologie, NARILIS, University of Namur (UNamur). Rue de Bruxelles 61, 5000

Unité de Recherche en Biologie des Microorganismes, Laboratoire d’Immunologie et de

10

Namur, Belgium.

11

@ Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB),

12

Gosselies, Belgium.

13

£ Laboratory of Neurophysiology, Université Libre de Bruxelles, Campus Erasme, Bldg. C,

14

808 Route de Lennik, 1070 Brussels, Belgium.

15

* Department of Molecular and Cellular Interactions, Vlaams Interuniversitair Instituut voor

16

Biotechnologie, Vrije Universiteit Brussel, Gebouw E, Verdieping 8, Pleinlaan 2, 1050

17

Brussels, Belgium.

18

† Laboratoire de Parasitologie. Faculté de Médecine. Route de Lennik 808, 1070 Bruxelles.

19

Université Libre de Bruxelles. Belgique.

20 21

1

Jean-Jacques Letesson and Eric Muraille are joint senior authors on this work.

22 23 24 25

Corresponding authors: - Eric Muraille. Mailing address: Laboratoire de Parasitologie. Faculté de médecine. Route de Lennik 808,

26

1070 Bruxelles. Université Libre de Bruxelles. Belgium. Fax.32 (0)2 5556128. Email:

27

[email protected]

28 29

Key words: Brucella melitensis, erythrocyte, infection

30 31

Running title: Erythrocyte infection by Brucella melitensis

32

1

33 34

ABSTRACT

35 36

Brucella spp are facultative intracellular Gram-negative coccobacilli responsible for

37

brucellosis, a worldwide zoonosis. We observed that Brucella melitensis is able to persist for

38

several weeks in the blood of intraperitoneally infected mice and transferred blood at any time

39

point tested is able to induce infection in naïve recipient mice. Bacterial persistence in the

40

blood is dramatically impaired by specific antibodies induced following Brucella vaccination.

41

In contrast to Bartonella, the Type IV secretion system and flagella expression are not

42

critically required for the persistence of Brucella in blood. ImageStream analysis of blood

43

cells showed that, following a brief extracellular phase, Brucella is mainly associated with the

44

erythrocytes. Examination by confocal microscopy and transmission electron microscopy

45

formally demonstrated that B. melitensis is able to invade erythrocytes in vivo. The bacteria

46

do not seem to multiply in erythrocytes and are found free in the cytoplasm. Our results open

47

up new areas for investigation and should serve in the development of novel strategies for the

48

treatment or prophylaxis of brucellosis. Invasion of erythrocytes could potentially protect the

49

bacterial cells from the host’s immune response, hamper antibiotic treatment and suggests

50

possible Brucella transmission by blood sucking insects in nature.

51 52 53

2

54

INTRODUCTION

55 56

Brucella is a facultative intracellular α-Proteobacteria that causes abortion and

57

infertility in mammals and leads to a debilitating febrile illness in humans. Although it is

58

rarely fatal, human brucellosis can progress into a severe chronic disease if left untreated (1,

59

2). Despite significant progress, the incidence of human brucellosis remains very high in

60

endemic areas (3). Complete eradication of Brucella would be unpractical due to its presence

61

in a large range of wild mammals (4, 5). Moreover, antibiotic treatment is costly and patients

62

frequently suffer from relapse of the bacteria (6, 7). Vaccination actually remains the only

63

rational strategy to confer protection on populations living in endemic countries.

64

Unfortunately, there is currently no available vaccine against human brucellosis as all

65

commercial animal vaccines are based on live attenuated strains of Brucella (8) that are still

66

virulent in humans. Efforts to develop an effective vaccine are impaired by our poor

67

knowledge of the Brucella life cycle in vivo and of the protective immune response induced

68

by infection. In particular, the mechanisms of early Brucella dissemination in vivo remain

69

largely unknown. A prominent characteristic of brucellosis is its long incubation period. The

70

current hypothesis is that this period, corresponding to reduced activation of innate immunity,

71

opens an “immunological window” and gives Brucella the chance to spread throughout the

72

organism to establish a replication niche within phagocytic cells (9).

73

Bacteremia is one major characteristic of human brucellosis (10) and is often

74

associated with an increased risk of relapse (6, 11). Moreover positive blood cultures are

75

synonymous with secondary seeding and development of focal complications of the disease.

76

Bacteremia in mice infected by intraperitoneal injection, the most frequent route of

77

experimental infection, has been reported in some studies (12, 13). However, to our

78

knowledge, the mechanism of Brucella persistence in the blood has never been investigated.

3

79

In this study, we monitored bacteremia in mice for several weeks after the intraperitoneal

80

inoculation of B. melitensis strain 16M constitutively expressing the mCherry fluorescent

81

tracer. The location of B. melitensis in the blood cells was analyzed by ImageStream analysis

82

and confocal microscopy. We also investigated the impact of type IV secretion system and

83

flagellum deficiencies as well as of specific antibodies on the blood persistence of the

84

bacteria.

85

Our results show that B. melitensis is able to persist for at least two weeks in the blood

86

of infected mice after intraperitoneal inoculation. The bacteria were first located

87

extracellularly but were then mainly found inside erythrocytes. To our knowledge, this

88

particular niche of infection has never been described in brucellosis, although it is relatively

89

common in other bacteria of the α-proteobacteria phylum such as Anaplasma (14) and

90

Bartonella (15). Circulating antibodies induced by vaccination drastically reduced blood

91

persistence of the bacteria following challenge.

92

4

MATERIAL AND METHODS

93 94 95

Ethics Statement

96

The animal handling and procedures used in this study complied with current

97

European legislation (directive 86/609/EEC) and the corresponding Belgian law “Arrêté royal

98

relatif à la protection des animaux d'expérience du 6 avril 2010 publié le 14 mai 2010"

99

(Royal Decree on the protection of experimental animals of April 6, 2010, published on May

100

14, 2010). The complete protocol was reviewed and approved by the Animal Welfare

101

Committee of the University of Namur (UNamur, Belgium) (Permit Number: 05-558).

102 103

Mice and reagents

104

MuMT−/− C57BL/6 mice (16) and C57BL/6 mice transgenic for GFP under the control

105

of the ubiquitin C promoter (Ubi-GFP) (17) were purchased from The Jackson Laboratory

106

(Bar Harbor, ME). Wild type C57BL/6 mice were purchased from Harlan (Bicester, UK) and

107

were used as the control. All wild type and deficient mice used in this study were bred in the

108

Gosselies animal facility of the Free University of Brussels (ULB, Belgium).

109

B. melitensis strain 16M (Biotype1, ATCC 23456) was grown in a biosafety level III

110

laboratory facility. Cultures grown overnight under shaking at 37°C in 2YT media (Luria-

111

Bertani broth with double quantity of yeast extract) to stationary phase were washed twice in

112

PBS (3500xg, 10 min.) as described previously (18). When indicated, we used B. melitensis

113

strain 16M which stably expresses the mCherry protein (mCherry-Br), a previously described

114

rapidly maturing variant of the red fluorescent protein DsRed (19), under the control of a

115

strong Brucella spp. promoter named PsojA (20). The construction of the mCherry-Br strain

116

has been described previously in detail (21). We also used the B. melitensis strain 16M

117

producing GFP (constructed by transferring the plasmid pBBR1-GFP-SOG (22) by mating),

5

118

the virB mutant B. melitensis strain 16M (deficient for the type IV secretion system, (23)) and

119

the fliC mutant B. melitensis strain 16M (deficient for flagellin expression, (24)).

120 121

Mouse infection

122

Mice were injected intraperitoneally (i.p.) with the indicated dose of B. melitensis in

123

500 μl of PBS. Control animals were injected with the same volume of PBS. The infectious

124

doses were validated by plating serial dilutions of inocula. Mice were bled at selected time

125

points and sacrificed at the end of the experiment.

126 127

Blood transfer experiment

128

Mice were injected i.p. (500 µl) with fresh blood from infected donor mice. Blood was

129

only diluted 20 fold in PBS before immediate administration. Two weeks later, spleens were

130

recovered in PBS/0.1% X-100 triton. Serial dilutions were performed in PBS and plated onto

131

2YT medium plates. The CFU counts were recorded after 4 days of culture at 37°C.

132 133

Antibiotic treatment

134

Mice were injected i.p. with 4x104 CFU of B. melitensis or with PBS. After resting for

135

3 weeks, antibiotic treatment was administered to both the immunized and control mice for 3

136

weeks. The treatment was a combination of rifampicin (12 mg/kg) and streptomycin (450

137

mg/kg) (adapted from (25)) prepared fresh daily and given in the drinking water. An

138

additional treatment was given i.p. and consisted of 5 injections of streptomycin (300 mg/kg)

139

throughout the 3 weeks of oral treatment.

140 141

6

142

RAW cell infection

143

RAW cells grown in 6-well tissue culture plates were inoculated with mCherry-Br at a

144

multiplicity of infection (MOI) of 300 in 2 ml of cell culture medium (DMEM 10% FBS).

145

Cells were then centrifuged at 1200 g for 10 minutes at 4°C and placed under 5% CO2 at

146

37°C. After 1 h, the cells were washed two times in PBS and incubated further with cell

147

culture medium supplemented with 50 μg/ml gentamicin until the end of the infection time.

148

After the indicated incubation time, the cells were washed three times in PBS, collected and

149

centrifuged at 1200 g for 10 minutes at 4°C. The cells were fixed in 300 μl of PBS/4% PFA

150

for further ImageStream analysis.

151 152

Bacterial count

153

For bacterial counts in the blood, 70 μl of blood were collected from the tail into

154

heparinized capillary tubes at the selected time points and diluted in PBS/0.1% X-100 triton

155

(Sigma). Serial dilutions were performed in PBS and plated onto 2YT media plates. CFUs

156

were counted after 4 days of culture at 37°C.

157

To separate the blood cells from the plasma, 140 μl of blood were collected and

158

diluted 4 times to allow for better separation. Samples were then centrifuged at 250g for 10

159

minutes at room temperature (RT) and each blood fraction was recovered. An additional

160

centrifugation (8000g, 2 minutes, RT) was performed on the supernatant of samples from

161

mice infected with a low dose of bacteria (4x104 CFU). This was necessary to concentrate the

162

bacteria. The supernatant fraction was diluted in PBS when necessary while the pellet fraction

163

was diluted in PBS/0.1% X-100 triton. Each fraction was then plated on 2YT and CFUs were

164

counted after 4 days of culture at 37°C.

165 166

7

Erythrocytes purification

167 168 169

Blood was collected from the tail into heparinized capillary tubes at the selected time

170

points, diluted in PBS and immediately filtered with Plasmodipur Filters (EuroProxima),

171

following the recommendation of the manufacturer. Plasmodipur Filters removes leukocytes

172

from blood (26, 27).

173 174

Cytofluorometric analysis

175

Blood was collected from the tail into heparinized capillary tubes at the selected time

176

points and diluted in PBS. After washing, blood cells were first incubated in saturating doses

177

of purified 2.4G2 (anti-mouse Fc receptor, ATCC) in 200 μl PBS 0.2% BSA 0.02% NaN3

178

(FACS buffer) for 20 minutes on ice to prevent antibody binding to Fc receptor. Blood cells

179

were stained on ice with various fluorescent mAb combinations in FACS buffer and further

180

collected on a FACScalibur cytofluorometer (Becton Dickinson, BD). We purchased the

181

following mAbs from BD Biosciences: Fluorescein (FITC)-coupled 145-2C11 (anti-CD3ε),

182

Phycoerythrin (PE)-coupled 1A8 (anti-Ly-6G) and Alexa647-coupled M5/114.15.2 (anti-

183

MHC-II).

184 185

Immunofluorescence microscopy on fixed bacteria

186

Blood from Ubiquitin-GFP C57BL/6 mice infected with 5x107 CFU of mCherry-Br

187

was diluted 1/1 (without preliminary wash or centrifugation) for 15 min in PBS containing

188

4% paraformaldehyde (pH 7.4) before being smeared on slides. The slides were dried

189

overnight at room temperature and then mounted in Fluoro-Gel medium containing DAPI

190

nucleic acid stain (Electron Microscopy Sciences, Hatfield, PA). The labeled cells were

191

visualized with an Axiovert M200 inverted microscope (Zeiss, Iena, Germany) equipped with

192

a high-resolution monochrome camera (AxioCam HR, Zeiss). Images (1384x1036 pixels

193

(0.16µm/pixel)) were acquired sequentially for each fluorochrome with an LD-Plan-NeoFluar

8

194

63x/0.75 N.A. dry objective and recorded as eight bit grey level zvi files. Images were

195

exported as TIFF files and figures were performed using Canvas 7 software.

196 197

Immunofluorescence microscopy on live bacteria

198

Wild type C57BL/6 mice were infected with 5x107 CFU of GFP or mCherry

199

expressing Brucella melitensis. Blood samples were harvested at 3h and 24h post infection

200

and immediately diluted 10x in PBS. For the observation, the samples were directly dropped

201

on an agarose pad (solution of 1% agarose in PBS), covered with a coverslip and sealed with

202

VALAP (1/3 Vaseline, 1/3 lanolin and 1/3 paraffin wax). The sealed pad was placed at 4°C

203

for 10 min before imaging with a Nikon 80i (objective Phase contrast 100X, plan Apo)

204

connected to a Hamamatsu ORCA-ER camera in a biosafety level III laboratory facility.

205 206

Multispectral Imaging Flow Cytometry (ImageStream)

207

Events were imaged using multispectral imaging flow cytometry (ImageStream 100,

208

Amnis Corp., Wash). At least 60,000 events were collected from each sample (100,000 for

209

blood samples) and the data were analyzed using image analysis software (IDEAS, Amnis

210

Corp., Wash). Bright field images of the cells and bacteria were recorded in channel 2 and

211

mCherry bacteria were recorded in channel 4. The acquisition gate included all cells and

212

bacteria and excluded fragments of cells and cell aggregates. Gate R1 was based on the

213

mCherry signal and allowed for the identification of bacteria inside and outside of blood cells.

214

The distinction between overlapping bacteria and extracellular (free or adherent) bacteria was

215

made by a combination of the erosion mask applied on the bright field image and the intensity

216

mask at 50% in the specific mCherry channel and using the intensity feature of IDEAS

217

software. Briefly, a bacterium was tagged as intracellular when its fluorescent image

9

218

overlapped with the bright field image of blood cells. The intracellular or extracellular

219

location of the bacteria was always confirmed by examination of the images.

220 221

Confocal microcopy

222

Confocal analyses were performed with the LSM780 confocal system fitted on an

223

Observer Z1 inverted microscope equipped with an alpha Plan Apochromat 63x/1.46 N.A. oil

224

immersion objective (Zeiss, Iena, Germany). The 488 nm excitation wavelength of the

225

Argon/2 laser, a main dichroic HFT 488 and a band-pass emission filter (BP500-530 nm)

226

were used for selective detection of the eGFP fluorochrome. The 543nm excitation

227

wavelength of the HeNe1 laser, a main dichroic HFT 488/543/633 and a long-pass emission

228

filter (BP580-650 nm) were used for selective detection of the mCherry fluorochrome. Stacks

229

of one micron-thick (i.e. 1 Airy Unit for the red fluorophore) optical sections (volume

230

sampled: 27 x 27 x 5.2 microns, 512 by 512 pixels, scaling: X & Y: 0.053 micron, Z: 0.370

231

micron) were collected sequentially for each fluorochrome throughout the thickness of the

232

object and stored as 16-bits *.lsm files. File Stacks were processed for deconvolution using

233

Huyghens® software (Scientific Volume Imaging BV, Hilversum, The Netherlands) and 3D

234

rendering was performed using Imaris® software (Bitplane AG, Zurich, Switzerland). Images

235

were exported as 8-bit uncompressed *.TIF image files.

236 237

Scanning electron microcopy

238

Samples were fixed overnight at 4°C in 4% PFA, 0.1M cacodylate buffer (pH 7.2),

239

and post-fixed in OsO4 (2%) in the same buffer. After serial dehydration, samples were dried

240

to the critical point and coated with platinum by standard procedures. Observations were

241

made in a Tecnai FEG ESEM QUANTA 200 (FEI) and images were processed by SIS iTEM

242

(Olympus) software.

10

243 244

Transmission electron microcopy

245

Infected red blood cells were fixed overnight in 2.5% glutaraldehyde in cacodylate

246

buffer, postfixed in 2% osmium tetroxide, serially dehydrated in ethanol and embedded in

247

epoxy resin (Agar 100 resin, Agar scientific, UK). Ultrathin 70 nm sections were obtained,

248

mounted on copper-formvar-carbon grids (EMS, UK) and stained with uranyl acetate and lead

249

citrate by standard procedures. Observations were made on a Tecnai 10 electron microscope

250

(FEI, Eindhoven) and images were captured with a Veleta CCD camera and processed with

251

AnalySIS and Adobe Photoshop software.

252 253

ELISA

254

Specific murine IgM, IgG1, IgG2a and IgG3 isotypes were determined by enzyme–

255

linked immunosorbent assay (ELISA). Polystyrene plates (Nunc 269620) were coated with

256

heat-killed B. melitensis (107 CFU/ml). After incubation overnight at 4°C, plates were

257

blocked for 2 hours at room temperature (RT) with 200 μl of PBS-3.65% casein. The plates

258

were then incubated for 1 h at RT with 50 μl of serial dilutions of the serum in PBS-3.5%

259

casein. Sera from naive mice were used as the negative control. After 4 washes with PBS,

260

isotype-specific goat anti-mouse horseradish peroxidase conjugates were added (50μl/well) at

261

appropriate dilutions (αIgM from Sigma; LO-MG1-13 HRPO, LO-MG2a-9 HRPO, LO-

262

MG3-13 HRPO from LO-IMEX). After 1 h of incubation at room temperature, the plates

263

were washed 4 times in PBS, and 100 μl/well of substrate solution (BD OptEiA) was added to

264

each well. After 10 minutes of incubation at room temperature in the dark, the enzyme

265

reaction was stopped by adding 25 μl/well of 2N H2SO4, and the absorbance was measured at

266

450 nm.

267

11

268

Statistical analysis

269

We used a (Wilcoxon-) Mann-Whitney test provided by the GraphPad Prism program

270

to statistically analyze our results. Each group of deficient mice was compared to wild type

271

mice. Values of p < 0.05 were considered to represent a significant difference. *, **, ***

272

denote p50% of the bacteria in the “72h blood”

464

group survived to the treatment with 200 µg/ml of gentamicin. In contrast, only around 10%

465

resisted in other groups (data not shown). Gentamicin is generally presented as an antibiotic

466

that only acts on extracellular bacteria but this is an oversimplification. Several studies have

467

demonstrated that gentamicin can kill intracellular bacteria inside macrophages (43). In

468

addition, pharmacokinetic studies have shown that gentamicin slowly penetrates human

469

erythrocytes (44). We suspect that infected erythrocytes may be more fragile and susceptible

470

to lyse during in vitro incubation. This can explain why bacteria, even when located inside

471

erythrocytes, remain partially sensible to gentamicin in our model.

472

In the model of erythrocyte infection by Bartonella spp, blood persistence and

473

erythrocyte invasion have been reported to be drastically reduced in absence of a functional

474

dependent type IV secretion system (29, 30) or flagella (31, 32). In our Brucella model of

475

infection, a bacterial strain deficient for flagella (FliC mutant) was able to persist in the blood

476

at a level that was similar to the wild type strain, thus demonstrating that flagella are not

477

implicated in blood persistence and presumably in erythrocyte invasion. Deficiency for the

478

type IV secretion system (VirB mutant) did not affect early persistence of bacteria in the

479

blood. Only blood CFU levels appeared to be reduced by 1 log at a later time point. As

480

persistence of the VirB mutant in the spleen is known to be reduced at a later time point (data

481

not shown and (23)), we can only conclude that the type IV secretion system is not critically

482

implicated in blood persistence as this strain remains able to persist until 13 days p.i., in

21

483

contrast to the complete inability of Bartonella type IV secretion system mutants to cause

484

bacteremia (29).

485

We also investigated whether the control of B. melitensis persistence in the blood

486

involves circulating antibodies and B cells. Some studies have reported that natural antibodies

487

play an important protective function in the control of early dissemination of viruses

488

(Vesicular Stomatitis Virus, Vaccinia virus, Lymphocytic ChorioMeningitis Virus) and

489

facultative intracellular bacteria (Listeria monocytogenes) (33, 34). Natural antibodies could

490

increase the early trapping of pathogens in secondary lymphoid organs by enhancing

491

phagocytosis. Previously, several groups have analyzed the role of natural antibodies in an

492

experimental model of Brucella, with contradictory results. Natural antibodies have been

493

reported to promote the control of an attenuated Brucella mutant by facilitating its elimination

494

by phagocytosis (45). Many serum transfer experiments have demonstrated that antibodies

495

play a positive role in protecting mice against Brucella infection (46-48). In this study, we

496

were unable to detect Brucella specific antibodies in naïve mice or during the 3 first days p.i.

497

Specific IgM antibodies were first detected at 6 days and IgG antibodies at 28 days, with a

498

predominance of the IgG2a- and IgG3 isotypes. This observation and the fact that the number

499

of bacteria found in the bloodstream during infection appears to be similar in wild type and

500

MuMT-/- mice demonstrated that natural antibodies had no or only a minor impact on

501

Brucella trapping in naïve mice under our experimental conditions. In contrast, we observed

502

that specific circulating antibodies present in vaccinated wild type mice dramatically reduced

503

the bacteria count persisting in the blood after a secondary infection. This suggests that the

504

blockage of Brucella erythrocyte colonization could constitute an important step in the

505

protective immune response against Brucella infection.

506

Although erythrocytes do not appear to constitute a replication niche for Brucella

507

since each infected cell contained only one single bacterium, they could serve as an important

22

508

part of the pathogen reservoir. Given the long lifespan of erythrocytes (49), Brucella could

509

potentially persist hidden in this environment for several months. The undulating fever

510

characteristic of human brucellosis and the high risk of relapse also argue for regularly

511

occurring new bacteremia waves. In this context, the failure to replicate within erythrocytes

512

could be advantageous for Brucella.

513

Of course, our data were obtained in the murine experimental model of infection with

514

Brucella melitensis and their relevance to other Brucella species and in the natural host

515

remains to be demonstrated. However, bacteremia is common in human brucellosis (11) and

516

an in vitro study has reported that Brucella is able to bind to sialic acid residues on human

517

erythrocytes (50), suggesting that erythrocyte invasion could also occur in patients.

518

From an evolutionary point of view, the unexpected ability of Brucella to infect

519

erythrocytes is not so surprising. Indeed, several bacterial pathogens belonging to the α-

520

(Bartonella spp (28), Anaplasma marginale (51)) and γ- (Francisella tularensis (52, 53))

521

proteobacteria phyla persist inside erythrocytes in the bloodstream during infection and are

522

transmitted to the host by ticks or fleas. Numerous old studies (5, 54-57) and a study

523

published in 2013 (58) have documented the presence of Brucella in blood-sucking

524

arthropods (ticks and fleas) under natural conditions. These observations, combined with our

525

demonstration of the ability of B. melitensis to infect erythrocytes in vivo, render plausible the

526

hypothesis that under natural conditions erythrocytes could constitute an alternative reservoir

527

for Brucella and that Brucella could be transmitted to the host by blood-sucking arthropods.

528

We hope that this hypothesis will be investigated in the future.

529

In conclusion, invasion of erythrocytes could constitute a crucial unexpected step that

530

promotes the evasion of host immune defenses. This new trait of Brucella pathogenesis opens

531

up new areas of research. Since erythrocytes are not capable of endocytosis (59), we will now

532

need to define the precise mechanism of Brucella uptake by the cell and identify the virulence

23

533

factors necessary for this process. We have tested several in vitro protocols to infect

534

erythrocytes from naïve mice. Unfortunately, under conditions where RAW cells were fully

535

infected, we never observed erythrocyte infection (data not shown). In addition, it would be

536

interesting to investigate the homing of infected erythrocytes in tissues and whether they

537

trigger a different immune response compared with other infected cells. We are thus left with

538

the exciting challenge of determining the importance of this mechanism to pathogenesis and

539

transmission of the infection.

540

24

541

ACKNOWLEDGEMENTS

542 543

This work was supported by grants from the Fonds National de la Recherche Scientifique

544

(FNRS) (FRSM FNRS convention 3.4.600.06.F, Belgium), from the Communauté Française

545

de Belgique (Action de Recherches Concertées 08/13–015), from the Interuniversity

546

Attraction

547

(http://www.belspo.be/belspo/iap/index_en.stm). E.M. is a Research Associate from the FRS-

548

FNRS (Belgium). M.A.V. and D.H.M hold PhD grants from the FRS-FNRS (Belgium). A.M.

549

and M.D. hold PhD grants from the FRIA (Belgium). The funding agencies played no role in

550

the design of the study, collection and analysis of the data, the decision to publish, or

551

preparation of the manuscript.

Poles

Programme

initiated

by

the

Belgian

Science

Policy

Office

552

25

FIGURE LEGENDS

553 554 555

Figure 1. Persistence of infectious B. melitensis in the blood of mice after i.p. infection.

556

A, B: Wild type C57BL/6 mice were injected i.p. with 4x104, 106 or 5x107 CFU of mCherry-

557

expressing B. melitensis (mCherry-Br) and blood was collected at selected time points. The

558

data represent either the mean +/- SEM of mCherry-Br CFU per ml of blood (A) or the

559

percentage of mice that still had bacteria in the blood (B). The results are representative of

560

two independent experiments. C, Wild type C57BL/6 mice were injected i.p. with 106 CFU of

561

mCherry-Br and blood was collected at 1, 3, 6 and 13 days p.i. Blood was immediately

562

diluted 20x in PBS and transferred by i.p. (500 µl) to recipient wild type C57BL/6 mice. Two

563

weeks after blood transfer, the mice were sacrificed and the spleens were harvested. The data

564

represent the CFU per spleen and are representative of two independent experiments. The

565

grey bars represent the medians.

566 567

Figure 2. Type IV secretion system (VirB) and flagella (FliC) deficient strains of B.

568

melitensis are able to persist in the blood of infected mice until 3 days. Wild type

569

C57BL/6 mice were injected i.p. with 106 CFU of wild type, FliC or VirB strain of B.

570

melitensis and blood was collected at the indicated time points. The data represent the CFU

571

per ml of blood of individual mice and are representative of three independent experiments.

572

The grey bars represent the medians.

573 574

Figure 3. Natural antibodies do not prevent B. melitensis dissemination in the blood

575

whereas vaccine-induced specific antibodies reduce the bacteremia following a

576

secondary infection. A, Wild type and MuMT-/- C57BL/6 mice were immunized i.p. with

577

4x104 CFU of B. melitensis (vaccinated group) or injected i.p. with PBS (naïve group) and

26

578

then treated with antibiotics for three weeks, as described in the Material and Methods

579

section. 60 days after immunization, all mice were injected i.p. with 5x107 CFU of mCherry-

580

Br and blood was collected at the indicated time points. The data represent the CFU per ml of

581

blood and are representative of three independent experiments. The grey bars represent the

582

medians. B, Wild-type C57BL/6 mice were infected with 4x104 CFU of B. melitensis. Serum

583

was collected at the indicated time points and ELISA was performed to determine the isotype

584

distribution of the Brucella-specific antibodies. The data represent the mean +/- SEM of 6

585

mice.

586 587

Figure 4. Blood location of B. melitensis after i.p. infection. A. Wild type C57BL/6 mice

588

were injected i.p. with 4x104 or 106 CFU of mCherry-Br and blood was collected at the

589

indicated time points. Samples were diluted and centrifuged to separate blood cells from

590

plasma. The data represent the mean +/- SD of the percentage of CFU in each blood fraction

591

and are representative of two independent experiments. B,C. Wild type C57BL/6 mice were

592

injected i.p. with 5x107 CFU of mCherry-Br and blood was collected at the indicated time

593

points. Samples were diluted and immediately filtered with plasmodipur filters. B. Total

594

blood cells and purified erythrocytes are analyzed by flow cytometry using anti-CD3ε, anti-

595

Ly-6G and anti-MHC-II antibodies in order to confirm the elimination of all leukocytes. C.

596

The data represent the CFU per ml of total blood or purified erythrocytes and are

597

representative of two independent experiments with 3 mice per group.

598 599

Figure 5. Immunofluorescence microscopy of fixed or live bacteria. A: Ubiquitin-GFP

600

C57BL/6 mice were infected i.p. with 5x107 CFU of mCherry-Br and bled at 3h and 24h p.i.

601

The blood was fixed and then smeared on slides. The slides were dried overnight and then

602

mounted in Fluoro-Gel medium containing DAPI nucleic acid stain (Electron Microscopy

27

603

Sciences, Hatfield, PA) and examined under a fluorescent microscope. Brucella was found

604

associated with erythrocytes mainly at 24h, and thus we present only images starting at 24h

605

p.i. The figure shows examples of mCherry bacteria associated with DAPI- erythrocytes. The

606

panels are color-coded according to the antigen examined. The scale bar = 20 or 10 µm, as

607

indicated. The data are representative of at least 3 independent experiments with a total of

608

more 200 bacteria observed. B, C: Wild type C57BL/6 mice were infected with 5x107 CFU of

609

mCherry-Br (B) or GFP-Br (C). Blood samples were harvested at 3h and 24h p.i. and

610

immediately diluted 10x in PBS. Freshly harvested blood samples were directly dropped on

611

an agarose pad and sealed with VALAP before observation under a fluorescent microscope in

612

a biosafety level III laboratory facility. The data are representative of at least 3 independent

613

experiments with a total of more than 30 bacteria observed under each condition.

614 615

Figure 6. ImageStream analysis of blood from infected mice

616

Wild type C57BL/6 Mice were injected i.p. with 5x107 CFU of mCherry-Br and blood was

617

collected at 3h, 24h and 72h p.i. The blood samples were immediately fixed by 3-fold dilution

618

in 4% paraformaldehyde and then analyzed by multispectral imaging flow cytometry

619

(ImageStream 100). As controls, we used a fresh culture of wild type Brucella, mCherry-Br

620

and a macrophage cell line (RAW cells) at 3h post in vitro infection with mCherry-Br. A.

621

Comparison of the percentage of events displaying a positive mCherry (channel 4) fluorescent

622

signal (gate R1) in blood from control and infected mice. B. Representative image of free

623

bacteria, uninfected and infected cells in channels 2 and 4. C, D. Kinetic analysis of the

624

percentage of free or adherent bacteria (in blue) and bacteria overlapping with cells (in green)

625

found in blood samples from control and infected mice. The location of bacteria was defined

626

by erosion mask. These data are representative of three independent experiments.

627

28

628

Figure 7. Brucella is found associated with or inside erythrocytes by scanning electron

629

and confocal microscopy. A. Scanning electron microscopy images from blood collected in

630

mice 3h p.i. by i.p. injection of 5x107 CFU of Brucella. Extracellular bacteria (a), attachment

631

of Brucella to erythrocytes (b), deformed erythrocytes suggesting Brucella infection (c-d). B.

632

Confocal microscopy images of blood collected in mice 24h p.i. by i.p. injection of 5x107

633

CFU of Brucella. Attachment of Brucella to erythrocytes (a), Brucella inside erythrocytes

634

(b).

635 636

Figure 8. Brucella is not surrounded by a vacuole in erythrocytes. Transmission electron

637

microscopy analysis of erythrocytes from mice 24h p.i. by i.p. injection of 5x107 CFU of

638

Brucella.

639 640 641 642

29

643

REFERENCES:

644 645 646 647 648

1.

Godfroid, J., A. Cloeckaert, J. P. Liautard, S. Kohler, D. Fretin, K. Walravens, B. Garin-Bastuji, and J. J. Letesson. 2005. From the discovery of the Malta fever's agent to the discovery of a marine mammal reservoir, brucellosis has continuously been a re-emerging zoonosis. Vet Res 36:313-326.

649 650

2.

Cutler, S. J., A. M. Whatmore, and N. J. Commander. 2005. Brucellosis--new aspects of an old disease. J Appl Microbiol 98:1270-1281.

651 652

3.

Pappas, G., P. Papadimitriou, N. Akritidis, L. Christou, and E. V. Tsianos. 2006. The new global map of human brucellosis. Lancet Infect Dis 6:91-99.

653 654 655

4.

Gregoire, F., B. Mousset, D. Hanrez, C. Michaux, K. Walravens, and A. Linden. 2012. A serological and bacteriological survey of brucellosis in wild boar (Sus scrofa) in Belgium. BMC Vet Res 8:80.

656 657

5.

Zheludkov, M. M., and L. E. Tsirel'son. 2010. Reservoirs of Brucella infection in nature. Biology Bulletin 37:709-715.

658 659

6.

Solera, J., E. Martinez-Alfaro, A. Espinosa, M. L. Castillejos, P. Geijo, and M. Rodriguez-Zapata. 1998. Multivariate model for predicting relapse in human brucellosis. J Infect 36:85-92.

660 661

7.

Ariza, J., J. Corredoira, R. Pallares, P. F. Viladrich, G. Rufi, M. Pujol, and F. Gudiol. 1995. Characteristics of and risk factors for relapse of brucellosis in humans. Clin Infect Dis 20:1241-1249.

662 663

8.

Schurig, G. G., N. Sriranganathan, and M. J. Corbel. 2002. Brucellosis vaccines: past, present and future. Vet Microbiol 90:479-496.

664 665

9.

Martirosyan, A., E. Moreno, and J. P. Gorvel. 2011. An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunol Rev 240:211-234.

666

10.

Yagupsky, P. 1999. Detection of Brucellae in blood cultures. J Clin Microbiol 37:3437-3442.

667 668

11.

Pappas, G., and P. Papadimitriou. 2007. Challenges in Brucella bacteraemia. Int J Antimicrob Agents 30 Suppl 1:S29-31.

669 670

12.

Rajashekara, G., D. A. Glover, M. Krepps, and G. A. Splitter. 2005. Temporal analysis of pathogenic events in virulent and avirulent Brucella melitensis infections. Cell Microbiol 7:1459-1473.

671 672

13.

Braude, A. I. 1951. Studies in the pathology and pathogenesis of experimental brucellosis. II. The formation of the hepatic granuloma and its evolution. J Infect Dis 89:87-94.

673 674

14.

Kocan, K. M., J. de la Fuente, A. A. Guglielmone, and R. D. Melendez. 2003. Antigens and alternatives for control of Anaplasma marginale infection in cattle. Clin Microbiol Rev 16:698-712.

675 676

15.

Harms, A., and C. Dehio. 2012. Intruders below the radar: molecular pathogenesis of Bartonella spp. Clin Microbiol Rev 25:42-78.

677 678

16.

Kitamura, D., J. Roes, R. Kuhn, and K. Rajewsky. 1991. A B cell-deficient mouse by targeted disruption of the membrane exon of the immunoglobulin mu chain gene. Nature 350:423-426.

679 680

17.

Schaefer, B. C., M. L. Schaefer, J. W. Kappler, P. Marrack, and R. M. Kedl. 2001. Observation of antigen-dependent CD8+ T-cell/ dendritic cell interactions in vivo. Cell Immunol 214:110-122.

30

681 682 683

18.

Copin, R., P. De Baetselier, Y. Carlier, J. J. Letesson, and E. Muraille. 2007. MyD88-Dependent Activation of B220-CD11b+LY-6C+ Dendritic Cells during Brucella melitensis Infection. J Immunol 178:5182-5191.

684 685 686

19.

Shaner, N. C., R. E. Campbell, P. A. Steinbach, B. N. Giepmans, A. E. Palmer, and R. Y. Tsien. 2004. Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat Biotechnol 22:1567-1572.

687 688

20.

Kohler, S., S. Ouahrani-Bettache, M. Layssac, J. Teyssier, and J. P. Liautard. 1999. Constitutive and inducible expression of green fluorescent protein in Brucella suis. Infect Immun 67:6695-6697.

689 690 691 692

21.

Copin, R., M. A. Vitry, D. Hanot Mambres, A. Machelart, C. De Trez, J. M. Vanderwinden, S. Magez, S. Akira, B. Ryffel, Y. Carlier, J. J. Letesson, and E. Muraille. 2012. In Situ Microscopy Analysis Reveals Local Innate Immune Response Developed around Brucella Infected Cells in Resistant and Susceptible Mice. PLoS Pathog 8:e1002575.

693 694

22.

Ouahrani-Bettache, S., F. Porte, J. Teyssier, J. P. Liautard, and S. Kohler. 1999. pBBR1-GFP: a broad-host-range vector for prokaryotic promoter studies. Biotechniques 26:620-622.

695 696 697

23.

Delrue, R. M., M. Martinez-Lorenzo, P. Lestrate, I. Danese, V. Bielarz, P. Mertens, X. De Bolle, A. Tibor, J. P. Gorvel, and J. J. Letesson. 2001. Identification of Brucella spp. genes involved in intracellular trafficking. Cell Microbiol 3:487-497.

698 699 700

24.

Leonard, S., J. Ferooz, V. Haine, I. Danese, D. Fretin, A. Tibor, S. de Walque, X. De Bolle, and J. J. Letesson. 2007. FtcR is a new master regulator of the flagellar system of Brucella melitensis 16M with homologs in Rhizobiaceae. J Bacteriol 189:131-141.

701 702 703

25.

Sathiyaseelan, J., R. Goenka, M. Parent, R. M. Benson, E. A. Murphy, D. M. Fernandes, A. S. Foulkes, and C. L. Baldwin. 2006. Treatment of Brucella-susceptible mice with IL-12 increases primary and secondary immunity. Cell Immunol 243:1-9.

704 705 706 707 708

26.

Auburn, S., S. Campino, T. G. Clark, A. A. Djimde, I. Zongo, R. Pinches, M. Manske, V. Mangano, D. Alcock, E. Anastasi, G. Maslen, B. Macinnis, K. Rockett, D. Modiano, C. I. Newbold, O. K. Doumbo, J. B. Ouedraogo, and D. P. Kwiatkowski. 2011. An effective method to purify Plasmodium falciparum DNA directly from clinical blood samples for whole genome highthroughput sequencing. PLoS One 6:e22213.

709 710 711

27.

Sriprawat, K., S. Kaewpongsri, R. Suwanarusk, M. L. Leimanis, U. Lek-Uthai, A. P. Phyo, G. Snounou, B. Russell, L. Renia, and F. Nosten. 2009. Effective and cheap removal of leukocytes and platelets from Plasmodium vivax infected blood. Malar J 8:115.

712 713

28.

Dehio, C. 2001. Bartonella interactions with endothelial cells and erythrocytes. Trends Microbiol 9:279-285.

714 715

29.

Schulein, R., and C. Dehio. 2002. The VirB/VirD4 type IV secretion system of Bartonella is essential for establishing intraerythrocytic infection. Mol Microbiol 46:1053-1067.

716 717 718 719

30.

Vayssier-Taussat, M., D. Le Rhun, H. K. Deng, F. Biville, S. Cescau, A. Danchin, G. Marignac, E. Lenaour, H. J. Boulouis, M. Mavris, L. Arnaud, H. Yang, J. Wang, M. Quebatte, P. Engel, H. Saenz, and C. Dehio. 2010. The Trw type IV secretion system of Bartonella mediates host-specific adhesion to erythrocytes. PLoS Pathog 6:e1000946.

720 721

31.

Benson, L. A., S. Kar, G. McLaughlin, and G. M. Ihler. 1986. Entry of Bartonella bacilliformis into erythrocytes. Infect Immun 54:347-353.

722 723 724

32.

Scherer, D. C., I. DeBuron-Connors, and M. F. Minnick. 1993. Characterization of Bartonella bacilliformis flagella and effect of antiflagellin antibodies on invasion of human erythrocytes. Infect Immun 61:4962-4971.

31

725 726 727

33.

Ochsenbein, A. F., T. Fehr, C. Lutz, M. Suter, F. Brombacher, H. Hengartner, and R. M. Zinkernagel. 1999. Control of early viral and bacterial distribution and disease by natural antibodies. Science 286:2156-2159.

728 729

34.

Boes, M. 2000. Role of natural and immune IgM antibodies in immune responses. Mol Immunol 37:1141-1149.

730 731 732

35.

Vitry, M. A., D. Hanot Mambres, C. De Trez, S. Akira, B. Ryffel, J. J. Letesson, and E. Muraille. 2014. Humoral Immunity and CD4+ Th1 Cells Are Both Necessary for a Fully Protective Immune Response upon Secondary Infection with Brucella melitensis. J Immunol 192.

733 734 735

36.

Billard, E., C. Cazevieille, J. Dornand, and A. Gross. 2005. High susceptibility of human dendritic cells to invasion by the intracellular pathogens Brucella suis, B. abortus, and B. melitensis. Infect Immun 73:8418-8424.

736 737 738

37.

Archambaud, C., S. P. Salcedo, H. Lelouard, E. Devilard, B. de Bovis, N. Van Rooijen, J. P. Gorvel, and B. Malissen. 2010. Contrasting roles of macrophages and dendritic cells in controlling initial pulmonary Brucella infection. Eur J Immunol 40:3458-3471.

739 740 741

38.

Enright, F. M., L. N. Araya, P. H. Elzer, G. E. Rowe, and A. J. Winter. 1990. Comparative histopathology in BALB/c mice infected with virulent and attenuated strains of Brucella abortus. Vet Immunol Immunopathol 26:171-182.

742 743

39.

Anderson, T. D., V. P. Meador, and N. F. Cheville. 1986. Pathogenesis of placentitis in the goat inoculated with Brucella abortus. I. Gross and histologic lesions. Vet Pathol 23:219-226.

744 745

40.

Detilleux, P. G., B. L. Deyoe, and N. F. Cheville. 1990. Penetration and intracellular growth of Brucella abortus in nonphagocytic cells in vitro. Infect Immun 58:2320-2328.

746 747 748

41.

Pizarro-Cerda, J., S. Meresse, R. G. Parton, G. van der Goot, A. Sola-Landa, I. Lopez-Goni, E. Moreno, and J. P. Gorvel. 1998. Brucella abortus transits through the autophagic pathway and replicates in the endoplasmic reticulum of nonprofessional phagocytes. Infect Immun 66:5711-5724.

749 750

42.

Holland, J. J., and M. J. Pickett. 1956. Intracellular behavior of Brucella variants in chick embryo cells in tissue culture. Proc Soc Exp Biol Med 93:476-479.

751 752

43.

Drevets, D. A., B. P. Canono, P. J. Leenen, and P. A. Campbell. 1994. Gentamicin kills intracellular Listeria monocytogenes. Infect Immun 62:2222-2228.

753 754 755

44.

Rolain, J. M., M. Maurin, M. N. Mallet, D. Parzy, and D. Raoult. 2003. Culture and antibiotic susceptibility of Bartonella quintana in human erythrocytes. Antimicrob Agents Chemother 47:614619.

756 757

45.

Rolan, H. G., M. N. Xavier, R. L. Santos, and R. M. Tsolis. 2009. Natural antibody contributes to host defense against an attenuated Brucella abortus virB mutant. Infect Immun 77:3004-3013.

758 759 760

46.

Winter, A. J., J. R. Duncan, C. G. Santisteban, J. T. Douglas, and L. G. Adams. 1989. Capacity of passively administered antibody to prevent establishment of Brucella abortus infection in mice. Infect Immun 57:3438-3444.

761 762 763

47.

Araya, L. N., P. H. Elzer, G. E. Rowe, F. M. Enright, and A. J. Winter. 1989. Temporal development of protective cell-mediated and humoral immunity in BALB/c mice infected with Brucella abortus. J Immunol 143:3330-3337.

764 765

48.

Montaraz, J. A., and A. J. Winter. 1986. Comparison of living and nonliving vaccines for Brucella abortus in BALB/c mice. Infect Immun 53:245-251.

766 767

49.

Shemin, D., and D. Rittenberg. 1946. The life span of the human red blood cell. J Biol Chem 166:627-636.

32

768 769 770

50.

del C. Rocha-Gracia, R., E. I. Castaneda-Roldan, S. Giono-Cerezo, and J. A. Giron. 2002. Brucella sp. bind to sialic acid residues on human and animal red blood cells. FEMS Microbiol Lett 213:219-224.

771 772 773

51.

de la Fuente, J., J. C. Garcia-Garcia, E. F. Blouin, J. T. Saliki, and K. M. Kocan. 2002. Infection of tick cells and bovine erythrocytes with one genotype of the intracellular ehrlichia Anaplasma marginale excludes infection with other genotypes. Clin Diagn Lab Immunol 9:658-668.

774 775 776

52.

Forestal, C. A., M. Malik, S. V. Catlett, A. G. Savitt, J. L. Benach, T. J. Sellati, and M. B. Furie. 2007. Francisella tularensis has a significant extracellular phase in infected mice. J Infect Dis 196:134137.

777 778

53.

Horzempa, J., D. M. O'Dee, D. B. Stolz, J. M. Franks, D. Clay, and G. J. Nau. 2011. Invasion of erythrocytes by Francisella tularensis. J Infect Dis 204:51-59.

779 780

54.

Wellmann, G. 1951. [Blood sucking insects as mechanical vectors of Brucella]. Zentralbl Bakteriol Orig 156:414-426.

781 782

55.

Wellmann, G. 1952. [Experimental studies on the possibility of Brucella transmission by insects]. Zentralbl Bakteriol Parasitenkd Infektionskr Hyg 159:71-86.

783 784

56.

Ozsan, K. 1962. [Isolation of Brucella melitensis from the fleas Xenopsylla conformis gathered from the burrows of wild rodents]. Ann Inst Pasteur (Paris) 103:90-92.

785 786

57.

Gudoshnik, A. N. 1958. [Role of pasture ticks and rodents in dissemination of Brucella]. Zh Mikrobiol Epidemiol Immunobiol 29:113-117.

787 788 789

58.

Neglia, G., V. Veneziano, E. De Carlo, G. Galiero, G. Borriello, M. Francillo, G. Campanile, L. Zicarelli, and L. Manna. 2013. Detection of Brucella abortus DNA and RNA in different stages of development of the sucking louse Haematopinus tuberculatus. BMC Vet Res 9:236.

790 791 792 793 794

59.

Schekman, R., and S. J. Singer. 1976. Clustering and endocytosis of membrane receptors can be induced in mature erythrocytes of neonatal but not adult humans. Proc Natl Acad Sci U S A 73:40754079.

795

33